751
|
Davey NE, Seo MH, Yadav VK, Jeon J, Nim S, Krystkowiak I, Blikstad C, Dong D, Markova N, Kim PM, Ivarsson Y. Discovery of short linear motif-mediated interactions through phage display of intrinsically disordered regions of the human proteome. FEBS J 2017; 284:485-498. [PMID: 28002650 DOI: 10.1111/febs.13995] [Citation(s) in RCA: 76] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Revised: 12/04/2016] [Accepted: 12/19/2016] [Indexed: 12/29/2022]
Abstract
The intrinsically disordered regions of eukaryotic proteomes are enriched in short linear motifs (SLiMs), which are of crucial relevance for cellular signaling and protein regulation; many mediate interactions by providing binding sites for peptide-binding domains. The vast majority of SLiMs remain to be discovered highlighting the need for experimental methods for their large-scale identification. We present a novel proteomic peptide phage display (ProP-PD) library that displays peptides representing the disordered regions of the human proteome, allowing direct large-scale interrogation of most potential binding SLiMs in the proteome. The performance of the ProP-PD library was validated through selections against SLiM-binding bait domains with distinct folds and binding preferences. The vast majority of identified binding peptides contained sequences that matched the known SLiM-binding specificities of the bait proteins. For SHANK1 PDZ, we establish a novel consensus TxF motif for its non-C-terminal ligands. The binding peptides mostly represented novel target proteins, however, several previously validated protein-protein interactions (PPIs) were also discovered. We determined the affinities between the VHS domain of GGA1 and three identified ligands to 40-130 μm through isothermal titration calorimetry, and confirmed interactions through coimmunoprecipitation using full-length proteins. Taken together, we outline a general pipeline for the design and construction of ProP-PD libraries and the analysis of ProP-PD-derived, SLiM-based PPIs. We demonstrated the methods potential to identify low affinity motif-mediated interactions for modular domains with distinct binding preferences. The approach is a highly useful complement to the current toolbox of methods for PPI discovery.
Collapse
Affiliation(s)
- Norman E Davey
- Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Ireland
| | - Moon-Hyeong Seo
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada
| | | | - Jouhyun Jeon
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada
| | - Satra Nim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada
| | - Izabella Krystkowiak
- Conway Institute of Biomolecular and Biomedical Sciences, University College Dublin, Ireland
| | | | - Debbie Dong
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada
| | | | - Philip M Kim
- Terrence Donnelly Centre for Cellular and Biomolecular Research, University of Toronto, Canada.,Department of Molecular Genetics and Department of Computer Science, University of Toronto, Canada
| | - Ylva Ivarsson
- Department of Chemistry - BMC, Uppsala University, Sweden
| |
Collapse
|
752
|
Pirone L, Xolalpa W, Sigurðsson JO, Ramirez J, Pérez C, González M, de Sabando AR, Elortza F, Rodriguez MS, Mayor U, Olsen JV, Barrio R, Sutherland JD. A comprehensive platform for the analysis of ubiquitin-like protein modifications using in vivo biotinylation. Sci Rep 2017; 7:40756. [PMID: 28098257 PMCID: PMC5241687 DOI: 10.1038/srep40756] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2016] [Accepted: 12/09/2016] [Indexed: 12/19/2022] Open
Abstract
Post-translational modification by ubiquitin and ubiquitin-like proteins (UbLs) is fundamental for maintaining protein homeostasis. Efficient isolation of UbL conjugates is hampered by multiple factors, including cost and specificity of reagents, removal of UbLs by proteases, distinguishing UbL conjugates from interactors, and low quantities of modified substrates. Here we describe bioUbLs, a comprehensive set of tools for studying modifications in Drosophila and mammals, based on multicistronic expression and in vivo biotinylation using the E. coli biotin protein ligase BirA. While the bioUbLs allow rapid validation of UbL conjugation for exogenous or endogenous proteins, the single vector approach can facilitate biotinylation of most proteins of interest. Purification under denaturing conditions inactivates deconjugating enzymes and stringent washes remove UbL interactors and non-specific background. We demonstrate the utility of the method in Drosophila cells and transgenic flies, identifying an extensive set of putative SUMOylated proteins in both cases. For mammalian cells, we show conjugation and localization for many different UbLs, with the identification of novel potential substrates for UFM1. Ease of use and the flexibility to modify existing vectors will make the bioUbL system a powerful complement to existing strategies for studying this important mode of protein regulation.
Collapse
Affiliation(s)
- Lucia Pirone
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Wendy Xolalpa
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Jón Otti Sigurðsson
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Juanma Ramirez
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain
| | - Coralia Pérez
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Monika González
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | | | - Félix Elortza
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - Manuel S Rodriguez
- ITAV, IPBS, Université de Toulouse, CNRS, UPS, 1 Place Pierre Potier Oncopole entrée B, BP 50624, 31106 Toulouse Cedex 1, France
| | - Ugo Mayor
- Biochemistry and Molecular Biology Department, University of the Basque Country (UPV/EHU), 48940 Leioa, Spain.,Ikerbasque, Basque Foundation for Science, Alameda Urquijo, 36-5 Plaza Bizkaia, 48011 Bilbao, Spain
| | - Jesper V Olsen
- Novo Nordisk Foundation Center for Protein Research, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3b, 2200 Copenhagen, Denmark
| | - Rosa Barrio
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| | - James D Sutherland
- CIC bioGUNE, Bizkaia Technology Park, Building 801-A, 48160 DERIO, Bizkaia, Spain
| |
Collapse
|
753
|
Kwon O, Kwak D, Ha SH, Jeon H, Park M, Chang Y, Suh PG, Ryu SH. Nudix-type motif 2 contributes to cancer proliferation through the regulation of Rag GTPase-mediated mammalian target of rapamycin complex 1 localization. Cell Signal 2017; 32:24-35. [PMID: 28089905 DOI: 10.1016/j.cellsig.2017.01.015] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2016] [Revised: 12/19/2016] [Accepted: 01/06/2017] [Indexed: 01/14/2023]
Abstract
Lysosomal localization of mammalian target of rapamycin complex 1 (mTORC1) is a critical step for activation of the molecule. Rag GTPases are essential for this translocation. Here, we demonstrate that Nudix-type motif 2 (NUDT2) is a novel positive regulator of mTORC1 activation. Activation of mTORC1 is impaired in NUDT2-silenced cells. Mechanistically, NUDT2 binds to Rag GTPase and controls mTORC1 translocation to the lysosomal membrane. Furthermore, NUDT2-dependent mTORC1 regulation is critical for proliferation of breast cancer cells, as NUDT2-silenced cells arrest in G0/G1 phases. Taken together, these results show that NUDT2 is a novel complex formation enhancing factor regulating mTORC1-Rag GTPase signaling that is crucial for cell growth control.
Collapse
Affiliation(s)
- Ohman Kwon
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Dongoh Kwak
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Sang Hoon Ha
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Hyeona Jeon
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Mangeun Park
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Yeonho Chang
- Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea
| | - Pann-Ghill Suh
- School of Nano-Biotechnology and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 689-798, Republic of Korea
| | - Sung Ho Ryu
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea; Department of Life Sciences, Pohang University of Science and Technology (POSTECH), Pohang 790-784, Republic of Korea.
| |
Collapse
|
754
|
Scott NE, Rogers LD, Prudova A, Brown NF, Fortelny N, Overall CM, Foster LJ. Interactome disassembly during apoptosis occurs independent of caspase cleavage. Mol Syst Biol 2017; 13:906. [PMID: 28082348 PMCID: PMC5293159 DOI: 10.15252/msb.20167067] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Protein-protein interaction networks (interactomes) define the functionality of all biological systems. In apoptosis, proteolysis by caspases is thought to initiate disassembly of protein complexes and cell death. Here we used a quantitative proteomics approach, protein correlation profiling (PCP), to explore changes in cytoplasmic and mitochondrial interactomes in response to apoptosis initiation as a function of caspase activity. We measured the response to initiation of Fas-mediated apoptosis in 17,991 interactions among 2,779 proteins, comprising the largest dynamic interactome to date. The majority of interactions were unaffected early in apoptosis, but multiple complexes containing known caspase targets were disassembled. Nonetheless, proteome-wide analysis of proteolytic processing by terminal amine isotopic labeling of substrates (TAILS) revealed little correlation between proteolytic and interactome changes. Our findings show that, in apoptosis, significant interactome alterations occur before and independently of caspase activity. Thus, apoptosis initiation includes a tight program of interactome rearrangement, leading to disassembly of relatively few, select complexes. These early interactome alterations occur independently of cleavage of these protein by caspases.
Collapse
Affiliation(s)
- Nichollas E Scott
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Lindsay D Rogers
- Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Anna Prudova
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Nat F Brown
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| | - Nikolaus Fortelny
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada.,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Christopher M Overall
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada.,Department of Oral Biological and Medical Sciences, University of British Columbia, Vancouver, BC, Canada.,Centre for Blood Research, University of British Columbia, Vancouver, BC, Canada
| | - Leonard J Foster
- Michael Smith Laboratories, University of British Columbia, Vancouver, BC, Canada .,Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
755
|
Wang S, Qu M, Peng J. PROSNET: INTEGRATING HOMOLOGY WITH MOLECULAR NETWORKS FOR PROTEIN FUNCTION PREDICTION. PACIFIC SYMPOSIUM ON BIOCOMPUTING. PACIFIC SYMPOSIUM ON BIOCOMPUTING 2017; 22:27-38. [PMID: 27896959 PMCID: PMC5319591 DOI: 10.1142/9789813207813_0004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Automated annotation of protein function has become a critical task in the post-genomic era. Network-based approaches and homology-based approaches have been widely used and recently tested in large-scale community-wide assessment experiments. It is natural to integrate network data with homology information to further improve the predictive performance. However, integrating these two heterogeneous, high-dimensional and noisy datasets is non-trivial. In this work, we introduce a novel protein function prediction algorithm ProSNet. An integrated heterogeneous network is first built to include molecular networks of multiple species and link together homologous proteins across multiple species. Based on this integrated network, a dimensionality reduction algorithm is introduced to obtain compact low-dimensional vectors to encode proteins in the network. Finally, we develop machine learning classification algorithms that take the vectors as input and make predictions by transferring annotations both within each species and across different species. Extensive experiments on five major species demonstrate that our integration of homology with molecular networks substantially improves the predictive performance over existing approaches.
Collapse
Affiliation(s)
- Sheng Wang
- Department of Computer Science, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | | | | |
Collapse
|
756
|
Mardakheh FK. Mass Spectrometry Analysis of Spatial Protein Networks by Colocalization Analysis (COLA). Methods Mol Biol 2017; 1636:337-352. [PMID: 28730490 DOI: 10.1007/978-1-4939-7154-1_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/11/2024]
Abstract
A major challenge in systems biology is comprehensive mapping of protein interaction networks. Crucially, such interactions are often dynamic in nature, necessitating methods that can rapidly mine the interactome across varied conditions and treatments to reveal change in the interaction networks. Recently, we described a fast mass spectrometry-based method to reveal functional interactions in mammalian cells on a global scale, by revealing spatial colocalizations between proteins (COLA) (Mardakheh et al., Mol Biosyst 13:92-105, 2017). As protein localization and function are inherently linked, significant colocalization between two proteins is a strong indication for their functional interaction. COLA uses rapid complete subcellular fractionation, coupled with quantitative proteomics to generate a subcellular localization profile for each protein quantified by the mass spectrometer. Robust clustering is then applied to reveal significant similarities in protein localization profiles, indicative of colocalization.
Collapse
Affiliation(s)
- Faraz K Mardakheh
- Centre for Molecular Oncology, Barts Cancer Institute, Queen Mary University of London, Charterhouse Square, London, EC1M 6BQ, UK.
| |
Collapse
|
757
|
Woodsmith J, Stelzl U, Vinayagam A. Bioinformatics Analysis of PTM-Modified Protein Interaction Networks and Complexes. Methods Mol Biol 2017; 1558:321-332. [PMID: 28150245 DOI: 10.1007/978-1-4939-6783-4_15] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Normal cellular functioning is maintained by macromolecular machines that control both core and specialized molecular tasks. These machines are in large part multi-subunit protein complexes that undergo regulation at multiple levels, from expression of requisite components to a vast array of post-translational modifications (PTMs). PTMs such as phosphorylation, ubiquitination, and acetylation currently number more than 200,000 in the human proteome and function within all molecular pathways. Here we provide a framework for systematically studying these PTMs in the context of global protein-protein interaction networks. This analytical framework allows insight into which functions specific PTMs tend to cluster in, and furthermore which complexes either single or multiple PTM signaling pathways converge on.
Collapse
Affiliation(s)
- Jonathan Woodsmith
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Ihnestrasse 63-73, Berlin, Germany
| | - Ulrich Stelzl
- Otto-Warburg Laboratory, Max-Planck Institute for Molecular Genetics (MPIMG), Ihnestrasse 63-73, Berlin, Germany.
- Department of Pharmaceutical Chemistry, Institute of Pharmaceutical Sciences, University of Graz, Universitätsplatz 1, Graz, Austria.
| | - Arunachalam Vinayagam
- Department of Genetics, Harvard Medical School, 77 Avenue Louis Pasteur, Boston, MA, 02115, USA
| |
Collapse
|
758
|
Yokoi M, Hanaoka F. Two mammalian homologs of yeast Rad23, HR23A and HR23B, as multifunctional proteins. Gene 2017; 597:1-9. [DOI: 10.1016/j.gene.2016.10.027] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 10/18/2016] [Indexed: 10/20/2022]
|
759
|
Exploring Protein-Protein Interactions as Drug Targets for Anti-cancer Therapy with In Silico Workflows. Methods Mol Biol 2017; 1647:221-236. [PMID: 28809006 DOI: 10.1007/978-1-4939-7201-2_15] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We describe a computational protocol to aid the design of small molecule and peptide drugs that target protein-protein interactions, particularly for anti-cancer therapy. To achieve this goal, we explore multiple strategies, including finding binding hot spots, incorporating chemical similarity and bioactivity data, and sampling similar binding sites from homologous protein complexes. We demonstrate how to combine existing interdisciplinary resources with examples of semi-automated workflows. Finally, we discuss several major problems, including the occurrence of drug-resistant mutations, drug promiscuity, and the design of dual-effect inhibitors.
Collapse
|
760
|
Jain KK. Personalized Management of Cardiovascular Disorders. Med Princ Pract 2017; 26:399-414. [PMID: 28898880 PMCID: PMC5757599 DOI: 10.1159/000481403] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2017] [Accepted: 09/11/2017] [Indexed: 12/28/2022] Open
Abstract
Personalized management of cardiovascular disorders (CVD), also referred to as personalized or precision cardiology in accordance with general principles of personalized medicine, is selection of the best treatment for an individual patient. It involves the integration of various "omics" technologies such as genomics and proteomics as well as other new technologies such as nanobiotechnology. Molecular diagnostics and biomarkers are important for linking diagnosis with therapy and monitoring therapy. Because CVD involve perturbations of large complex biological networks, a systems biology approach to CVD risk stratification may be used for improving risk-estimating algorithms, and modeling of personalized benefit of treatment may be helpful for guiding the choice of intervention. Bioinformatics tools are helpful in analyzing and integrating large amounts of data from various sources. Personalized therapy is considered during drug development, including methods of targeted drug delivery and clinical trials. Individualized recommendations consider multiple factors - genetic as well as epigenetic - for patients' risk of heart disease. Examples of personalized treatment are those of chronic myocardial ischemia, heart failure, and hypertension. Similar approaches can be used for the management of atrial fibrillation and hypercholesterolemia, as well as the use of anticoagulants. Personalized management includes pharmacotherapy, surgery, lifestyle modifications, and combinations thereof. Further progress in understanding the pathomechanism of complex cardiovascular diseases and identification of causative factors at the individual patient level will provide opportunities for the development of personalized cardiology. Application of principles of personalized medicine will improve the care of the patients with CVD.
Collapse
Affiliation(s)
- Kewal K. Jain
- *Prof. K.K. Jain, MD, FRACS, FFPM, CEO, Jain PharmaBiotech, Bläsiring 7, CH-4057 Basel (Switzerland), E-Mail
| |
Collapse
|
761
|
Salazar BM, Balczewski EA, Ung CY, Zhu S. Neuroblastoma, a Paradigm for Big Data Science in Pediatric Oncology. Int J Mol Sci 2016; 18:E37. [PMID: 28035989 PMCID: PMC5297672 DOI: 10.3390/ijms18010037] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Revised: 12/14/2016] [Accepted: 12/17/2016] [Indexed: 12/13/2022] Open
Abstract
Pediatric cancers rarely exhibit recurrent mutational events when compared to most adult cancers. This poses a challenge in understanding how cancers initiate, progress, and metastasize in early childhood. Also, due to limited detected driver mutations, it is difficult to benchmark key genes for drug development. In this review, we use neuroblastoma, a pediatric solid tumor of neural crest origin, as a paradigm for exploring "big data" applications in pediatric oncology. Computational strategies derived from big data science-network- and machine learning-based modeling and drug repositioning-hold the promise of shedding new light on the molecular mechanisms driving neuroblastoma pathogenesis and identifying potential therapeutics to combat this devastating disease. These strategies integrate robust data input, from genomic and transcriptomic studies, clinical data, and in vivo and in vitro experimental models specific to neuroblastoma and other types of cancers that closely mimic its biological characteristics. We discuss contexts in which "big data" and computational approaches, especially network-based modeling, may advance neuroblastoma research, describe currently available data and resources, and propose future models of strategic data collection and analyses for neuroblastoma and other related diseases.
Collapse
Affiliation(s)
- Brittany M Salazar
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA.
| | - Emily A Balczewski
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | - Choong Yong Ung
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| | - Shizhen Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine, Rochester, MN 55902, USA.
- Department of Molecular Pharmacology and Experimental Therapeutics, Mayo Clinic College of Medicine, Rochester, MN 55905, USA.
| |
Collapse
|
762
|
Yu L, Wang B, Ma X, Gao L. The extraction of drug-disease correlations based on module distance in incomplete human interactome. BMC SYSTEMS BIOLOGY 2016; 10:111. [PMID: 28155709 PMCID: PMC5260043 DOI: 10.1186/s12918-016-0364-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Abstract
BACKGROUND Extracting drug-disease correlations is crucial in unveiling disease mechanisms, as well as discovering new indications of available drugs, or drug repositioning. Both the interactome and the knowledge of disease-associated and drug-associated genes remain incomplete. RESULTS We present a new method to predict the associations between drugs and diseases. Our method is based on a module distance, which is originally proposed to calculate distances between modules in incomplete human interactome. We first map all the disease genes and drug genes to a combined protein interaction network. Then based on the module distance, we calculate the distances between drug gene sets and disease gene sets, and take the distances as the relationships of drug-disease pairs. We also filter possible false positive drug-disease correlations by p-value. Finally, we validate the top-100 drug-disease associations related to six drugs in the predicted results. CONCLUSION The overlapping between our predicted correlations with those reported in Comparative Toxicogenomics Database (CTD) and literatures, and their enriched Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways demonstrate our approach can not only effectively identify new drug indications, but also provide new insight into drug-disease discovery.
Collapse
Affiliation(s)
- Liang Yu
- School of Computer Science and Technology, Xidian University, Xi’an, 710071 People’s Republic of China
| | - Bingbo Wang
- School of Computer Science and Technology, Xidian University, Xi’an, 710071 People’s Republic of China
| | - Xiaoke Ma
- School of Computer Science and Technology, Xidian University, Xi’an, 710071 People’s Republic of China
| | - Lin Gao
- School of Computer Science and Technology, Xidian University, Xi’an, 710071 People’s Republic of China
| |
Collapse
|
763
|
Hill SM, Nesser NK, Johnson-Camacho K, Jeffress M, Johnson A, Boniface C, Spencer SEF, Lu Y, Heiser LM, Lawrence Y, Pande NT, Korkola JE, Gray JW, Mills GB, Mukherjee S, Spellman PT. Context Specificity in Causal Signaling Networks Revealed by Phosphoprotein Profiling. Cell Syst 2016; 4:73-83.e10. [PMID: 28017544 PMCID: PMC5279869 DOI: 10.1016/j.cels.2016.11.013] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2015] [Revised: 08/06/2016] [Accepted: 11/23/2016] [Indexed: 01/08/2023]
Abstract
Signaling networks downstream of receptor tyrosine kinases are among the most extensively studied biological networks, but new approaches are needed to elucidate causal relationships between network components and understand how such relationships are influenced by biological context and disease. Here, we investigate the context specificity of signaling networks within a causal conceptual framework using reverse-phase protein array time-course assays and network analysis approaches. We focus on a well-defined set of signaling proteins profiled under inhibition with five kinase inhibitors in 32 contexts: four breast cancer cell lines (MCF7, UACC812, BT20, and BT549) under eight stimulus conditions. The data, spanning multiple pathways and comprising ∼70,000 phosphoprotein and ∼260,000 protein measurements, provide a wealth of testable, context-specific hypotheses, several of which we experimentally validate. Furthermore, the data provide a unique resource for computational methods development, permitting empirical assessment of causal network learning in a complex, mammalian setting. Time-course assays of signaling proteins in cancer cell lines under kinase inhibition Causal conceptual framework for network analysis Data shed light on causal protein networks that are specific to biological context Resource for signaling biology and for benchmarking computational methods
Collapse
Affiliation(s)
- Steven M Hill
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK
| | - Nicole K Nesser
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97201, USA
| | - Katie Johnson-Camacho
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97201, USA
| | | | - Aimee Johnson
- Bayer Healthcare North America, Berkeley, CA 94710, USA
| | - Chris Boniface
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97201, USA
| | - Simon E F Spencer
- Department of Statistics, University of Warwick, Coventry CV4 7AL, UK
| | - Yiling Lu
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97201, USA
| | - Yancey Lawrence
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97201, USA
| | - Nupur T Pande
- Department of Obstetrics and Gynecology, Women's Health Research Unit, Oregon Health and Science University, Portland, OR 97239, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA
| | - James E Korkola
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97201, USA
| | - Joe W Gray
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97201, USA; Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97239, USA; Center for Spatial Systems Biomedicine, Oregon Health and Science University, Portland, OR 97239, USA
| | - Gordon B Mills
- Department of Systems Biology, MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Sach Mukherjee
- MRC Biostatistics Unit, University of Cambridge, Cambridge CB2 0SR, UK; German Center for Neurodegenerative Diseases (DZNE), Bonn 53127, Germany.
| | - Paul T Spellman
- Department of Molecular and Medical Genetics, Oregon Health and Science University, Portland, OR 97201, USA.
| |
Collapse
|
764
|
Mardakheh FK, Sailem HZ, Kümper S, Tape CJ, McCully RR, Paul A, Anjomani-Virmouni S, Jørgensen C, Poulogiannis G, Marshall CJ, Bakal C. Proteomics profiling of interactome dynamics by colocalisation analysis (COLA). MOLECULAR BIOSYSTEMS 2016; 13:92-105. [PMID: 27824369 PMCID: PMC5315029 DOI: 10.1039/c6mb00701e] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/12/2016] [Accepted: 11/01/2016] [Indexed: 12/27/2022]
Abstract
Localisation and protein function are intimately linked in eukaryotes, as proteins are localised to specific compartments where they come into proximity of other functionally relevant proteins. Significant co-localisation of two proteins can therefore be indicative of their functional association. We here present COLA, a proteomics based strategy coupled with a bioinformatics framework to detect protein-protein co-localisations on a global scale. COLA reveals functional interactions by matching proteins with significant similarity in their subcellular localisation signatures. The rapid nature of COLA allows mapping of interactome dynamics across different conditions or treatments with high precision.
Collapse
Affiliation(s)
- Faraz K Mardakheh
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| | - Heba Z Sailem
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK. and Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford OX3 7DQ, UK
| | - Sandra Kümper
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| | - Christopher J Tape
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK. and Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, USA
| | - Ryan R McCully
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| | - Angela Paul
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| | - Sara Anjomani-Virmouni
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| | - Claus Jørgensen
- Cancer Research UK Manchester Institute, University of Manchester, Wilmslow Road, Manchester M20 4BX, UK
| | - George Poulogiannis
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| | - Christopher J Marshall
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| | - Chris Bakal
- Institute of Cancer Research, Division of Cancer Biology, 237 Fulham Road, London SW3 6JB, UK.
| |
Collapse
|
765
|
Kafková L, Debler EW, Fisk JC, Jain K, Clarke SG, Read LK. The Major Protein Arginine Methyltransferase in Trypanosoma brucei Functions as an Enzyme-Prozyme Complex. J Biol Chem 2016; 292:2089-2100. [PMID: 27998975 DOI: 10.1074/jbc.m116.757112] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/14/2016] [Indexed: 11/06/2022] Open
Abstract
Prozymes are catalytically inactive enzyme paralogs that dramatically stimulate the function of weakly active enzymes through complex formation. The two prozymes described to date reside in the polyamine biosynthesis pathway of the human parasite Trypanosoma brucei, an early branching eukaryote that lacks transcriptional regulation and regulates its proteome through posttranscriptional and posttranslational means. Arginine methylation is a common posttranslational modification in eukaryotes catalyzed by protein arginine methyltransferases (PRMTs) that are typically thought to function as homodimers. We demonstrate that a major T. brucei PRMT, TbPRMT1, functions as a heterotetrameric enzyme-prozyme pair. The inactive PRMT paralog, TbPRMT1PRO, is essential for catalytic activity of the TbPRMT1ENZ subunit. Mutational analysis definitively demonstrates that TbPRMT1ENZ is the cofactor-binding subunit and carries all catalytic activity of the complex. Our results are the first demonstration of an obligate heteromeric PRMT, and they suggest that enzyme-prozyme organization is expanded in trypanosomes as a posttranslational means of enzyme regulation.
Collapse
Affiliation(s)
- Lucie Kafková
- From the Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, and Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14214
| | - Erik W Debler
- the Laboratory of Cell Biology, The Rockefeller University, New York, New York 10065, and
| | - John C Fisk
- From the Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, and Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14214
| | - Kanishk Jain
- the Department of Chemistry and Biochemistry and The Molecular Biology Institute, UCLA, Los Angeles, California 90095
| | - Steven G Clarke
- the Department of Chemistry and Biochemistry and The Molecular Biology Institute, UCLA, Los Angeles, California 90095
| | - Laurie K Read
- From the Department of Microbiology and Immunology, Witebsky Center for Microbial Pathogenesis and Immunology, and Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, New York 14214,
| |
Collapse
|
766
|
Forero DA, Prada CF, Perry G. Functional and Genomic Features of Human Genes Mutated in Neuropsychiatric Disorders. Open Neurol J 2016; 10:143-148. [PMID: 27990183 PMCID: PMC5120378 DOI: 10.2174/1874205x01610010143] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Revised: 09/08/2016] [Accepted: 09/16/2016] [Indexed: 11/22/2022] Open
Abstract
Background: In recent years, a large number of studies around the world have led to the identification of causal genes for hereditary types of common and rare neurological and psychiatric disorders. Objective: To explore the functional and genomic features of known human genes mutated in neuropsychiatric disorders. Methods: A systematic search was used to develop a comprehensive catalog of genes mutated in neuropsychiatric disorders (NPD). Functional enrichment and protein-protein interaction analyses were carried out. A false discovery rate approach was used for correction for multiple testing. Results: We found several functional categories that are enriched among NPD genes, such as gene ontologies, protein domains, tissue expression, signaling pathways and regulation by brain-expressed miRNAs and transcription factors. Sixty six of those NPD genes are known to be druggable. Several topographic parameters of protein-protein interaction networks and the degree of conservation between orthologous genes were identified as significant among NPD genes. Conclusion: These results represent one of the first analyses of enrichment of functional categories of genes known to harbor mutations for NPD. These findings could be useful for a future creation of computational tools for prioritization of novel candidate genes for NPD.
Collapse
Affiliation(s)
- Diego A Forero
- Laboratory of NeuroPsychiatric Genetics, Biomedical Sciences Research Group, School of Medicine, Universidad Antonio Nariño, Bogotá, Colombia
| | - Carlos F Prada
- Grupo de Citogenética, Filogenia y Evolución de Poblaciones, Universidad del Tolima. Ibagué, Colombia
| | - George Perry
- College of Sciences, University of Texas at San Antonio, San Antonio, Texas, USA
| |
Collapse
|
767
|
Salivary and pellicle proteome: A datamining analysis. Sci Rep 2016; 6:38882. [PMID: 27966577 PMCID: PMC5155218 DOI: 10.1038/srep38882] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 11/16/2016] [Indexed: 01/06/2023] Open
Abstract
We aimed to comprehensively compare two compartmented oral proteomes, the salivary and the dental pellicle proteome. Systematic review and datamining was used to obtain the physico-chemical, structural, functional and interactional properties of 1,515 salivary and 60 identified pellicle proteins. Salivary and pellicle proteins did not differ significantly in their aliphatic index, hydrophaty, instability index, or isoelectric point. Pellicle proteins were significantly more charged at low and high pH and were significantly smaller (10–20 kDa) than salivary proteins. Protein structure and solvent accessible molecular surface did not differ significantly. Proteins of the pellicle were more phosphorylated and glycosylated than salivary proteins. Ion binding and enzymatic activities also differed significantly. Protein-protein-ligand interaction networks relied on few key proteins. The identified differences between salivary and pellicle proteins could guide proteome compartmentalization and result in specialized functionality. Key proteins could be potential targets for diagnostic or therapeutic application.
Collapse
|
768
|
Chatr-Aryamontri A, Oughtred R, Boucher L, Rust J, Chang C, Kolas NK, O'Donnell L, Oster S, Theesfeld C, Sellam A, Stark C, Breitkreutz BJ, Dolinski K, Tyers M. The BioGRID interaction database: 2017 update. Nucleic Acids Res 2016; 45:D369-D379. [PMID: 27980099 PMCID: PMC5210573 DOI: 10.1093/nar/gkw1102] [Citation(s) in RCA: 702] [Impact Index Per Article: 78.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 10/25/2016] [Accepted: 10/27/2016] [Indexed: 01/05/2023] Open
Abstract
The Biological General Repository for Interaction Datasets (BioGRID: https://thebiogrid.org) is an open access database dedicated to the annotation and archival of protein, genetic and chemical interactions for all major model organism species and humans. As of September 2016 (build 3.4.140), the BioGRID contains 1 072 173 genetic and protein interactions, and 38 559 post-translational modifications, as manually annotated from 48 114 publications. This dataset represents interaction records for 66 model organisms and represents a 30% increase compared to the previous 2015 BioGRID update. BioGRID curates the biomedical literature for major model organism species, including humans, with a recent emphasis on central biological processes and specific human diseases. To facilitate network-based approaches to drug discovery, BioGRID now incorporates 27 501 chemical-protein interactions for human drug targets, as drawn from the DrugBank database. A new dynamic interaction network viewer allows the easy navigation and filtering of all genetic and protein interaction data, as well as for bioactive compounds and their established targets. BioGRID data are directly downloadable without restriction in a variety of standardized formats and are freely distributed through partner model organism databases and meta-databases.
Collapse
Affiliation(s)
- Andrew Chatr-Aryamontri
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3T 1J4, Canada
| | - Rose Oughtred
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Lorrie Boucher
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Jennifer Rust
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Christie Chang
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Nadine K Kolas
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Lara O'Donnell
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Sara Oster
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Chandra Theesfeld
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Adnane Sellam
- Centre Hospitalier de l'Université Laval (CHUL), Québec, Québec G1V 4G2, Canada
| | - Chris Stark
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Bobby-Joe Breitkreutz
- The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| | - Kara Dolinski
- Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Mike Tyers
- Institute for Research in Immunology and Cancer, Université de Montréal, Montréal, Quebec H3T 1J4, Canada .,The Lunenfeld-Tanenbaum Research Institute, Mount Sinai Hospital, Toronto, Ontario M5G 1X5, Canada
| |
Collapse
|
769
|
Jadot M, Boonen M, Thirion J, Wang N, Xing J, Zhao C, Tannous A, Qian M, Zheng H, Everett JK, Moore DF, Sleat DE, Lobel P. Accounting for Protein Subcellular Localization: A Compartmental Map of the Rat Liver Proteome. Mol Cell Proteomics 2016; 16:194-212. [PMID: 27923875 DOI: 10.1074/mcp.m116.064527] [Citation(s) in RCA: 58] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 11/18/2016] [Indexed: 11/06/2022] Open
Abstract
Accurate knowledge of the intracellular location of proteins is important for numerous areas of biomedical research including assessing fidelity of putative protein-protein interactions, modeling cellular processes at a system-wide level and investigating metabolic and disease pathways. Many proteins have not been localized, or have been incompletely localized, partly because most studies do not account for entire subcellular distribution. Thus, proteins are frequently assigned to one organelle whereas a significant fraction may reside elsewhere. As a step toward a comprehensive cellular map, we used subcellular fractionation with classic balance sheet analysis and isobaric labeling/quantitative mass spectrometry to assign locations to >6000 rat liver proteins. We provide quantitative data and error estimates describing the distribution of each protein among the eight major cellular compartments: nucleus, mitochondria, lysosomes, peroxisomes, endoplasmic reticulum, Golgi, plasma membrane and cytosol. Accounting for total intracellular distribution improves quality of organelle assignments and assigns proteins with multiple locations. Protein assignments and supporting data are available online through the Prolocate website (http://prolocate.cabm.rutgers.edu). As an example of the utility of this data set, we have used organelle assignments to help analyze whole exome sequencing data from an infant dying at 6 months of age from a suspected neurodegenerative lysosomal storage disorder of unknown etiology. Sequencing data was prioritized using lists of lysosomal proteins comprising well-established residents of this organelle as well as novel candidates identified in this study. The latter included copper transporter 1, encoded by SLC31A1, which we localized to both the plasma membrane and lysosome. The patient harbors two predicted loss of function mutations in SLC31A1, suggesting that this may represent a heretofore undescribed recessive lysosomal storage disease gene.
Collapse
Affiliation(s)
- Michel Jadot
- From the ‡URPhyM-Laboratoire de Chimie Physiologique, Université de Namur, 61 rue de Bruxelles, Namur 5000, Belgium;
| | - Marielle Boonen
- From the ‡URPhyM-Laboratoire de Chimie Physiologique, Université de Namur, 61 rue de Bruxelles, Namur 5000, Belgium
| | - Jaqueline Thirion
- From the ‡URPhyM-Laboratoire de Chimie Physiologique, Université de Namur, 61 rue de Bruxelles, Namur 5000, Belgium
| | - Nan Wang
- §Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Jinchuan Xing
- §Department of Genetics, Human Genetics Institute of New Jersey, Rutgers, The State University of New Jersey, Piscataway, NJ 08854
| | - Caifeng Zhao
- ¶Center for Advanced Biotechnology and Medicine, Rutgers Biomedical and Health Sciences, 679 Hoes Lane West, Piscataway, New Jersey 08854
| | - Abla Tannous
- ¶Center for Advanced Biotechnology and Medicine, Rutgers Biomedical and Health Sciences, 679 Hoes Lane West, Piscataway, New Jersey 08854
| | - Meiqian Qian
- ¶Center for Advanced Biotechnology and Medicine, Rutgers Biomedical and Health Sciences, 679 Hoes Lane West, Piscataway, New Jersey 08854
| | - Haiyan Zheng
- ¶Center for Advanced Biotechnology and Medicine, Rutgers Biomedical and Health Sciences, 679 Hoes Lane West, Piscataway, New Jersey 08854
| | - John K Everett
- ¶Center for Advanced Biotechnology and Medicine, Rutgers Biomedical and Health Sciences, 679 Hoes Lane West, Piscataway, New Jersey 08854
| | - Dirk F Moore
- ‖Department of Biostatistics, School of Public Health, Rutgers Biomedical and Health Sciences, 683 Hoes Lane West, Piscataway, New Jersey 08854
| | - David E Sleat
- ¶Center for Advanced Biotechnology and Medicine, Rutgers Biomedical and Health Sciences, 679 Hoes Lane West, Piscataway, New Jersey 08854;
| | - Peter Lobel
- ¶Center for Advanced Biotechnology and Medicine, Rutgers Biomedical and Health Sciences, 679 Hoes Lane West, Piscataway, New Jersey 08854;
| |
Collapse
|
770
|
Global vision of druggability issues: applications and perspectives. Drug Discov Today 2016; 22:404-415. [PMID: 27939283 DOI: 10.1016/j.drudis.2016.11.021] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 10/10/2016] [Accepted: 11/25/2016] [Indexed: 02/04/2023]
Abstract
During the preliminary stage of a drug discovery project, the lack of druggability information and poor target selection are the main causes of frequent failures. Elaborating on accurate computational druggability prediction methods is a requirement for prioritizing target selection, designing new drugs and avoiding side effects. In this review, we describe a survey of recently reported druggability prediction methods mainly based on networks, statistical pocket druggability predictions and virtual screening. An application for a frequent mutation of p53 tumor suppressor is presented, illustrating the complementarity of druggability prediction approaches, the remaining challenges and potential new drug development perspectives.
Collapse
|
771
|
Yang W, Bang H, Jang K, Sung MK, Choi JK. Predicting the recurrence of noncoding regulatory mutations in cancer. BMC Bioinformatics 2016; 17:492. [PMID: 27912731 PMCID: PMC5135808 DOI: 10.1186/s12859-016-1385-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2016] [Accepted: 11/26/2016] [Indexed: 11/25/2022] Open
Abstract
Background One of the greatest challenges in cancer genomics is to distinguish driver mutations from passenger mutations. Whereas recurrence is a hallmark of driver mutations, it is difficult to observe recurring noncoding mutations owing to a limited amount of whole-genome sequenced samples. Hence, it is required to develop a method to predict potentially recurrent mutations. Results In this work, we developed a random forest classifier that predicts regulatory mutations that may recur based on the features of the mutations repeatedly appearing in a given cohort. With breast cancer as a model, we profiled 35 quantitative features describing genetic and epigenetic signals at the mutation site, transcription factors whose binding motif was disrupted by the mutation, and genes targeted by long-range chromatin interactions. A true set of mutations for machine learning was generated by interrogating publicly available pan-cancer genomes based on our statistical model of mutation recurrence. The performance of our random forest classifier was evaluated by cross validations. The variable importance of each feature in the classification of mutations was investigated. Our statistical recurrence model for the random forest classifier showed an area under the curve (AUC) of ~0.78 in predicting recurrent mutations. Chromatin accessibility at the mutation sites, the distance from the mutations to known cancer risk loci, and the role of the target genes in the regulatory or protein interaction network were among the most important variables. Conclusions Our methods enable to characterize recurrent regulatory mutations using a limited number of whole-genome samples, and based on the characterization, to predict potential driver mutations whose recurrence is not found in the given samples but likely to be observed with additional samples. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1385-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Woojin Yang
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Hyoeun Bang
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Kiwon Jang
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Min Kyung Sung
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea
| | - Jung Kyoon Choi
- Department of Bio and Brain Engineering, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
772
|
Goh WWB, Wong L. Integrating Networks and Proteomics: Moving Forward. Trends Biotechnol 2016; 34:951-959. [DOI: 10.1016/j.tibtech.2016.05.015] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Revised: 05/23/2016] [Accepted: 05/24/2016] [Indexed: 11/28/2022]
|
773
|
Expanding the Immunology Toolbox: Embracing Public-Data Reuse and Crowdsourcing. Immunity 2016; 45:1191-1204. [DOI: 10.1016/j.immuni.2016.12.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Revised: 11/30/2016] [Accepted: 12/01/2016] [Indexed: 12/15/2022]
|
774
|
Guo Y, An L, Ng HM, Sy SMH, Huen MSY. An E2-guided E3 Screen Identifies the RNF17-UBE2U Pair as Regulator of the Radiosensitivity, Immunodeficiency, Dysmorphic Features, and Learning Difficulties (RIDDLE) Syndrome Protein RNF168. J Biol Chem 2016; 292:967-978. [PMID: 27903633 DOI: 10.1074/jbc.m116.758854] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2016] [Revised: 11/14/2016] [Indexed: 12/22/2022] Open
Abstract
Protein ubiquitination has emerged as a pivotal regulatory reaction that promotes cellular responses to DNA damage. With a goal to delineate the DNA damage signal transduction cascade, we systematically analyzed the human E2 ubiquitin- and ubiquitin-like-conjugating enzymes for their ability to mobilize the DNA damage marker 53BP1 onto ionizing radiation-induced DNA double strand breaks. An RNAi-based screen identified UBE2U as a candidate regulator of chromatin responses at double strand breaks. Further mining of the UBE2U interactome uncovered its cognate E3 RNF17 as a novel factor that, via the radiosensitivity, immunodeficiency, dysmorphic features, and learning difficulties (RIDDLE) syndrome protein RNF168, enforces DNA damage responses. Our screen allowed us to uncover new players in the mammalian DNA damage response and highlights the instrumental roles of ubiquitin machineries in promoting cell responses to genotoxic stress.
Collapse
Affiliation(s)
- Yingying Guo
- From the School of Biomedical Sciences, Li Ka Shing Faculty of Medicine and
| | - Liwei An
- From the School of Biomedical Sciences, Li Ka Shing Faculty of Medicine and
| | - Hoi-Man Ng
- From the School of Biomedical Sciences, Li Ka Shing Faculty of Medicine and
| | - Shirley M H Sy
- From the School of Biomedical Sciences, Li Ka Shing Faculty of Medicine and
| | - Michael S Y Huen
- From the School of Biomedical Sciences, Li Ka Shing Faculty of Medicine and .,the State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong, China
| |
Collapse
|
775
|
Estruch SB, Graham SA, Chinnappa SM, Deriziotis P, Fisher SE. Functional characterization of rare FOXP2 variants in neurodevelopmental disorder. J Neurodev Disord 2016; 8:44. [PMID: 27933109 PMCID: PMC5126810 DOI: 10.1186/s11689-016-9177-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2016] [Accepted: 11/08/2016] [Indexed: 01/15/2023] Open
Abstract
Background Heterozygous disruption of FOXP2 causes a rare form of speech and language impairment. Screens of the FOXP2 sequence in individuals with speech/language-related disorders have identified several rare protein-altering variants, but their phenotypic relevance is often unclear. FOXP2 encodes a transcription factor with a forkhead box DNA-binding domain, but little is known about the functions of protein regions outside this domain. Methods We performed detailed functional analyses of seven rare FOXP2 variants found in affected cases, including three which have not been previously characterized, testing intracellular localization, transcriptional regulation, dimerization, and interaction with other proteins. To shed further light on molecular functions of FOXP2, we characterized the interaction between this transcription factor and co-repressor proteins of the C-terminal binding protein (CTBP) family. Finally, we analysed the functional significance of the polyglutamine tracts in FOXP2, since tract length variations have been reported in cases of neurodevelopmental disorder. Results We confirmed etiological roles of multiple FOXP2 variants. Of three variants that have been suggested to cause speech/language disorder, but never before been characterized, only one showed functional effects. For the other two, we found no effects on protein function in any assays, suggesting that they are incidental to the phenotype. We identified a CTBP-binding region within the N-terminal portion of FOXP2. This region includes two amino acid substitutions that occurred on the human lineage following the split from chimpanzees. However, we did not observe any effects of these amino acid changes on CTBP binding or other core aspects of FOXP2 function. Finally, we found that FOXP2 variants with reduced polyglutamine tracts did not exhibit altered behaviour in cellular assays, indicating that such tracts are non-essential for core aspects of FOXP2 function, and that tract variation is unlikely to be a highly penetrant cause of speech/language disorder. Conclusions Our findings highlight the importance of functional characterization of novel rare variants in FOXP2 in assessing the contribution of such variants to speech/language disorder and provide further insights into the molecular function of the FOXP2 protein. Electronic supplementary material The online version of this article (doi:10.1186/s11689-016-9177-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Sara B Estruch
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, the Netherlands
| | - Sarah A Graham
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, the Netherlands
| | - Swathi M Chinnappa
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, the Netherlands
| | - Pelagia Deriziotis
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, the Netherlands
| | - Simon E Fisher
- Language and Genetics Department, Max Planck Institute for Psycholinguistics, Wundtlaan 1, 6525 XD Nijmegen, the Netherlands.,Donders Institute for Brain, Cognition and Behaviour, Radboud University, 6525 EN Nijmegen, the Netherlands
| |
Collapse
|
776
|
Wang CY, Shahi P, Huang JTW, Phan NN, Sun Z, Lin YC, Lai MD, Werb Z. Systematic analysis of the achaete-scute complex-like gene signature in clinical cancer patients. Mol Clin Oncol 2016; 6:7-18. [PMID: 28123722 PMCID: PMC5244854 DOI: 10.3892/mco.2016.1094] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 09/23/2016] [Indexed: 12/11/2022] Open
Abstract
The achaete-scute complex-like (ASCL) family, also referred to as ‘achaete-scute complex homolog’ or ‘achaete-scute family basic helix-loop-helix transcription factor’, is critical for proper development of the nervous system and deregulation of ASCL plays a key role in psychiatric and neurological disorders. The ASCL family consists of five members, namely ASCL1, ASCL2, ASCL3, ASCL4 and ASCL5. The ASCL1 gene serves as a potential oncogene during lung cancer development. There is a correlation between increased ASCL2 expression and colon cancer development. Inhibition of ASCL2 reduced cellular proliferation and tumor growth in xenograft tumor experiments. Although previous studies demonstrated involvement of ASCL1 and ASCL2 in tumor development, little is known on the remaining ASCL family members and their potential effect on tumorigenesis. Therefore, a holistic approach to investigating the expression of ASCL family genes in diverse types of cancer may provide new insights in cancer research. In this study, we utilized a web-based microarray database (Oncomine; www.oncomine.org) to analyze the transcriptional expression of the ASCL family in clinical cancer and normal tissues. Our bioinformatics analysis revealed the potential involvement of multiple ASCL family members during tumor onset and progression in multiple types of cancer. Compared to normal tissue, ASCL1 exhibited a higher expression in cancers of the lung, pancreas, kidney, esophagus and head and neck, whereas ASCL2 exhibited a high expression in cancers of the breast, colon, stomach, lung, head and neck, ovary and testis. ASCL3, however, exhibited a high expression only in breast cancer. Interestingly, ASCL1 expression was downregulated in melanoma and in cancers of the bladder, breast, stomach and colon. ASCL2 exhibited low expression levels in sarcoma, melanoma, brain and prostate cancers. Reduction in the expression of ASCL3 was detected in lymphoma, bladder, cervical, kidney and epithelial cancers. Similarly, ASCL5 exhibited low expression in the majority of brain cancer subtypes, such as glioblastoma and oligodendroglioma. This analysis supports the hypothesis that specific ASCL members may play an important role in cancer development. Collectively, our data suggest that alterations in the expression of ASCL gene family members are correlated with cancer development. Furthermore, ASCL family members were categorized according to cancer subtype. The aim of this report was to provide novel insights to the significance of the ASCL family in various cancers and our findings suggested that the ASCL gene family may be an ideal target for future cancer studies.
Collapse
Affiliation(s)
- Chih-Yang Wang
- Department of Anatomy, University of California, San Francisco, CA 94143, USA; Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan 11114, R.O.C.; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan 11114, R.O.C
| | - Payam Shahi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA 94143, USA
| | - John Ting Wei Huang
- Department of Oncology, University of California, San Francisco, CA 94143, USA
| | - Nam Nhut Phan
- Faculty of Applied Sciences, Ton Duc Thang University, Ho Chi Minh 7000, Vietnam; Graduate Institute of Biotechnology, Chinese Culture University, Taipei, Taiwan 11114, R.O.C
| | - Zhengda Sun
- Department of Radiology, University of California, San Francisco, CA 94143, USA
| | - Yen-Chang Lin
- Graduate Institute of Biotechnology, Chinese Culture University, Taipei, Taiwan 11114, R.O.C
| | - Ming-Derg Lai
- Department of Biochemistry and Molecular Biology, College of Medicine, National Cheng Kung University, Tainan, Taiwan 11114, R.O.C.; Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan 11114, R.O.C
| | - Zena Werb
- Department of Anatomy, University of California, San Francisco, CA 94143, USA
| |
Collapse
|
777
|
Gomez-Cabrero D, Menche J, Vargas C, Cano I, Maier D, Barabási AL, Tegnér J, Roca J. From comorbidities of chronic obstructive pulmonary disease to identification of shared molecular mechanisms by data integration. BMC Bioinformatics 2016; 17:441. [PMID: 28185567 PMCID: PMC5133493 DOI: 10.1186/s12859-016-1291-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Background Deep mining of healthcare data has provided maps of comorbidity relationships between diseases. In parallel, integrative multi-omics investigations have generated high-resolution molecular maps of putative relevance for understanding disease initiation and progression. Yet, it is unclear how to advance an observation of comorbidity relations (one disease to others) to a molecular understanding of the driver processes and associated biomarkers. Results Since Chronic Obstructive Pulmonary disease (COPD) has emerged as a central hub in temporal comorbidity networks, we developed a systematic integrative data-driven framework to identify shared disease-associated genes and pathways, as a proxy for the underlying generative mechanisms inducing comorbidity. We integrated records from approximately 13 M patients from the Medicare database with disease-gene maps that we derived from several resources including a semantic-derived knowledge-base. Using rank-based statistics we not only recovered known comorbidities but also discovered a novel association between COPD and digestive diseases. Furthermore, our analysis provides the first set of COPD co-morbidity candidate biomarkers, including IL15, TNF and JUP, and characterizes their association to aging and life-style conditions, such as smoking and physical activity. Conclusions The developed framework provides novel insights in COPD and especially COPD co-morbidity associated mechanisms. The methodology could be used to discover and decipher the molecular underpinning of other comorbidity relationships and furthermore, allow the identification of candidate co-morbidity biomarkers. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1291-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- David Gomez-Cabrero
- Department of Medicine, Karolinska Institutet, Unit of Computational Medicine, Stockholm, 171 77, Sweden. .,Karolinska Institutet, Center for Molecular Medicine, Stockholm, 171 77, Sweden. .,Department of Medicine, Unit of Clinical Epidemiology, Karolinska University Hospital, Solna, L8, 17176, Sweden. .,Science for Life Laboratory, Solna, 17121, Sweden. .,Mucosal and Salivary Biology Division, King's College London Dental Institute, London, UK.
| | - Jörg Menche
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Center for Network Science, Central European University, Budapest, Hungary
| | - Claudia Vargas
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic de Barcelona, Universitat de Barcelona, Barcelona, Spain.,Center for Biomedical Network Research in Respiratory Diseases (CIBERES), Madrid, Spain
| | - Isaac Cano
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic de Barcelona, Universitat de Barcelona, Barcelona, Spain.,Center for Biomedical Network Research in Respiratory Diseases (CIBERES), Madrid, Spain
| | | | - Albert-László Barabási
- Center for Complex Networks Research and Department of Physics, Northeastern University, Boston, MA, USA.,Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.,Center for Network Science, Central European University, Budapest, Hungary.,Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Jesper Tegnér
- Department of Medicine, Karolinska Institutet, Unit of Computational Medicine, Stockholm, 171 77, Sweden.,Karolinska Institutet, Center for Molecular Medicine, Stockholm, 171 77, Sweden.,Department of Medicine, Unit of Clinical Epidemiology, Karolinska University Hospital, Solna, L8, 17176, Sweden.,Science for Life Laboratory, Solna, 17121, Sweden
| | - Josep Roca
- Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic de Barcelona, Universitat de Barcelona, Barcelona, Spain. .,Center for Biomedical Network Research in Respiratory Diseases (CIBERES), Madrid, Spain.
| | | |
Collapse
|
778
|
Jinawath N, Bunbanjerdsuk S, Chayanupatkul M, Ngamphaiboon N, Asavapanumas N, Svasti J, Charoensawan V. Bridging the gap between clinicians and systems biologists: from network biology to translational biomedical research. J Transl Med 2016; 14:324. [PMID: 27876057 PMCID: PMC5120462 DOI: 10.1186/s12967-016-1078-3] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 11/08/2016] [Indexed: 01/22/2023] Open
Abstract
With the wealth of data accumulated from completely sequenced genomes and other high-throughput experiments, global studies of biological systems, by simultaneously investigating multiple biological entities (e.g. genes, transcripts, proteins), has become a routine. Network representation is frequently used to capture the presence of these molecules as well as their relationship. Network biology has been widely used in molecular biology and genetics, where several network properties have been shown to be functionally important. Here, we discuss how such methodology can be useful to translational biomedical research, where scientists traditionally focus on one or a small set of genes, diseases, and drug candidates at any one time. We first give an overview of network representation frequently used in biology: what nodes and edges represent, and review its application in preclinical research to date. Using cancer as an example, we review how network biology can facilitate system-wide approaches to identify targeted small molecule inhibitors. These types of inhibitors have the potential to be more specific, resulting in high efficacy treatments with less side effects, compared to the conventional treatments such as chemotherapy. Global analysis may provide better insight into the overall picture of human diseases, as well as identify previously overlooked problems, leading to rapid advances in medicine. From the clinicians’ point of view, it is necessary to bridge the gap between theoretical network biology and practical biomedical research, in order to improve the diagnosis, prevention, and treatment of the world’s major diseases.
Collapse
Affiliation(s)
- Natini Jinawath
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand.,Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Sacarin Bunbanjerdsuk
- Program in Translational Medicine, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Maneerat Chayanupatkul
- Department of Physiology, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand.,Division of Gastroenterology and Hepatology, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - Nuttapong Ngamphaiboon
- Medical Oncology Unit, Department of Medicine Faculty of Medicine, Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Nithi Asavapanumas
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Jisnuson Svasti
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand.,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand.,Laboratory of Biochemistry, Chulabhorn Research Institute, Bangkok, Thailand
| | - Varodom Charoensawan
- Integrative Computational BioScience (ICBS) Center, Mahidol University, Nakhon Pathom, Thailand. .,Department of Biochemistry, Faculty of Science, Mahidol University, Bangkok, Thailand. .,Systems Biology of Diseases Research Unit, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
779
|
Horton ER, Humphries JD, James J, Jones MC, Askari JA, Humphries MJ. The integrin adhesome network at a glance. J Cell Sci 2016; 129:4159-4163. [PMID: 27799358 PMCID: PMC5117201 DOI: 10.1242/jcs.192054] [Citation(s) in RCA: 132] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The adhesion nexus is the site at which integrin receptors bridge intracellular cytoskeletal and extracellular matrix networks. The connection between integrins and the cytoskeleton is mediated by a dynamic integrin adhesion complex (IAC), the components of which transduce chemical and mechanical signals to control a multitude of cellular functions. In this Cell Science at a Glance article and the accompanying poster, we integrate the consensus adhesome, a set of 60 proteins that have been most commonly identified in isolated IAC proteomes, with the literature-curated adhesome, a theoretical network that has been assembled through scholarly analysis of proteins that localise to IACs. The resulting IAC network, which comprises four broad signalling and actin-bridging axes, provides a platform for future studies of the regulation and function of the adhesion nexus in health and disease.
Collapse
Affiliation(s)
- Edward R Horton
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Jonathan D Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Jenny James
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Matthew C Jones
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Janet A Askari
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| | - Martin J Humphries
- Wellcome Trust Centre for Cell-Matrix Research, Faculty of Biology, Medicine and Health, University of Manchester, Manchester M13 9PT, UK
| |
Collapse
|
780
|
Pellegrini M, Baglioni M, Geraci F. Protein complex prediction for large protein protein interaction networks with the Core&Peel method. BMC Bioinformatics 2016; 17:372. [PMID: 28185552 PMCID: PMC5123419 DOI: 10.1186/s12859-016-1191-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023] Open
Abstract
Background Biological networks play an increasingly important role in the exploration of functional modularity and cellular organization at a systemic level. Quite often the first tools used to analyze these networks are clustering algorithms. We concentrate here on the specific task of predicting protein complexes (PC) in large protein-protein interaction networks (PPIN). Currently, many state-of-the-art algorithms work well for networks of small or moderate size. However, their performance on much larger networks, which are becoming increasingly common in modern proteome-wise studies, needs to be re-assessed. Results and discussion We present a new fast algorithm for clustering large sparse networks: Core&Peel, which runs essentially in time and storage O(a(G)m+n) for a network G of n nodes and m arcs, where a(G) is the arboricity of G (which is roughly proportional to the maximum average degree of any induced subgraph in G). We evaluated Core&Peel on five PPI networks of large size and one of medium size from both yeast and homo sapiens, comparing its performance against those of ten state-of-the-art methods. We demonstrate that Core&Peel consistently outperforms the ten competitors in its ability to identify known protein complexes and in the functional coherence of its predictions. Our method is remarkably robust, being quite insensible to the injection of random interactions. Core&Peel is also empirically efficient attaining the second best running time over large networks among the tested algorithms. Conclusions Our algorithm Core&Peel pushes forward the state-of the-art in PPIN clustering providing an algorithmic solution with polynomial running time that attains experimentally demonstrable good output quality and speed on challenging large real networks. Electronic supplementary material The online version of this article (doi:10.1186/s12859-016-1191-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marco Pellegrini
- Laboratory for Integrative Systems Medicine - Istituto di Informatica e Telematica and Istituto di Fisiologia Clinica del CNR, via Moruzzi 1, Pisa, 56124, Italy.
| | - Miriam Baglioni
- Laboratory for Integrative Systems Medicine - Istituto di Informatica e Telematica and Istituto di Fisiologia Clinica del CNR, via Moruzzi 1, Pisa, 56124, Italy
| | - Filippo Geraci
- Laboratory for Integrative Systems Medicine - Istituto di Informatica e Telematica and Istituto di Fisiologia Clinica del CNR, via Moruzzi 1, Pisa, 56124, Italy
| |
Collapse
|
781
|
Folador EL, de Carvalho PVSD, Silva WM, Ferreira RS, Silva A, Gromiha M, Ghosh P, Barh D, Azevedo V, Röttger R. In silico identification of essential proteins in Corynebacterium pseudotuberculosis based on protein-protein interaction networks. BMC SYSTEMS BIOLOGY 2016; 10:103. [PMID: 27814699 PMCID: PMC5097352 DOI: 10.1186/s12918-016-0346-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 10/18/2016] [Indexed: 12/27/2022]
Abstract
Background Corynebacterium pseudotuberculosis (Cp) is a gram-positive bacterium that is classified into equi and ovis serovars. The serovar ovis is the etiological agent of caseous lymphadenitis, a chronic infection affecting sheep and goats, causing economic losses due to carcass condemnation and decreased production of meat, wool, and milk. Current diagnosis or treatment protocols are not fully effective and, thus, require further research of Cp pathogenesis. Results Here, we mapped known protein-protein interactions (PPI) from various species to nine Cp strains to reconstruct parts of the potential Cp interactome and to identify potentially essential proteins serving as putative drug targets. On average, we predict 16,669 interactions for each of the nine strains (with 15,495 interactions shared among all strains). An in silico sanity check suggests that the potential networks were not formed by spurious interactions but have a strong biological bias. With the inferred Cp networks we identify 181 essential proteins, among which 41 are non-host homologous. Conclusions The list of candidate interactions of the Cp strains lay the basis for developing novel hypotheses and designing according wet-lab studies. The non-host homologous essential proteins are attractive targets for therapeutic and diagnostic proposes. They allow for searching of small molecule inhibitors of binding interactions enabling modern drug discovery. Overall, the predicted Cp PPI networks form a valuable and versatile tool for researchers interested in Corynebacterium pseudotuberculosis. Electronic supplementary material The online version of this article (doi:10.1186/s12918-016-0346-4) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Edson Luiz Folador
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Institute of Biological Sciences, Federal University of Para, Belém, PA, Brazil.,Biotechnology Center (CBiotec), Federal University of Paraiba (UFPB), João Pessoa, Brazil
| | - Paulo Vinícius Sanches Daltro de Carvalho
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil.,Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark
| | - Wanderson Marques Silva
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Rafaela Salgado Ferreira
- Department of Biochemistry and Immunology, Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Artur Silva
- Institute of Biological Sciences, Federal University of Para, Belém, PA, Brazil
| | - Michael Gromiha
- Department of Biotechnology, Indian Institute of Technology (IIT) Madras, Tamilnadu, India
| | - Preetam Ghosh
- Department of Computer Science, Virginia Commonwealth University, Richmond, VA, USA
| | - Debmalya Barh
- Centre for Genomics and Applied Gene Technology, Institute of Integrative Omics and Applied Biotechnology (IIOAB), Nonakuri, Purba Medinipur, West Bengal, India
| | - Vasco Azevedo
- Department of General Biology, Instituto de Ciências Biológicas (ICB), Federal University of Minas Gerais (UFMG), Belo Horizonte, Brazil
| | - Richard Röttger
- Department of Mathematics and Computer Science, University of Southern Denmark, Odense, Denmark.
| |
Collapse
|
782
|
Pulido-Tamayo S, Weytjens B, De Maeyer D, Marchal K. SSA-ME Detection of cancer driver genes using mutual exclusivity by small subnetwork analysis. Sci Rep 2016; 6:36257. [PMID: 27808240 PMCID: PMC5093737 DOI: 10.1038/srep36257] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2015] [Accepted: 10/11/2016] [Indexed: 11/21/2022] Open
Abstract
Because of its clonal evolution a tumor rarely contains multiple genomic alterations in the same pathway as disrupting the pathway by one gene often is sufficient to confer the complete fitness advantage. As a result, many cancer driver genes display mutual exclusivity across tumors. However, searching for mutually exclusive gene sets requires analyzing all possible combinations of genes, leading to a problem which is typically too computationally complex to be solved without a stringent a priori filtering, restricting the mutations included in the analysis. To overcome this problem, we present SSA-ME, a network-based method to detect cancer driver genes based on independently scoring small subnetworks for mutual exclusivity using a reinforced learning approach. Because of the algorithmic efficiency, no stringent upfront filtering is required. Analysis of TCGA cancer datasets illustrates the added value of SSA-ME: well-known recurrently mutated but also rarely mutated drivers are prioritized. We show that using mutual exclusivity to detect cancer driver genes is complementary to state-of-the-art approaches. This framework, in which a large number of small subnetworks are being analyzed in order to solve a computationally complex problem (SSA), can be generically applied to any problem in which local neighborhoods in a network hold useful information.
Collapse
Affiliation(s)
- Sergio Pulido-Tamayo
- Department of Information Technology, iGent Toren, Technologiepark 15, 9052 Gent, Belgium.,Department of Plant Biotechnology and Bioinformatics, UGent, Technologiepark 927, 9052 Gent, Belgium.,Bioinformatics Institute Ghent, Technologiepark 927, 9052 Gent, Belgium.,Dept. of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium.,Grupo de Investigación en Ciencias Biológicas y Bioprocesos (Cibiop), Universidad EAFIT, Carrera 49 N° 7 Sur-50, Medellín, Colombia
| | - Bram Weytjens
- Department of Information Technology, iGent Toren, Technologiepark 15, 9052 Gent, Belgium.,Department of Plant Biotechnology and Bioinformatics, UGent, Technologiepark 927, 9052 Gent, Belgium.,Bioinformatics Institute Ghent, Technologiepark 927, 9052 Gent, Belgium.,Dept. of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| | - Dries De Maeyer
- Department of Information Technology, iGent Toren, Technologiepark 15, 9052 Gent, Belgium.,Department of Plant Biotechnology and Bioinformatics, UGent, Technologiepark 927, 9052 Gent, Belgium.,Bioinformatics Institute Ghent, Technologiepark 927, 9052 Gent, Belgium.,Dept. of Microbial and Molecular Systems, KU Leuven, Kasteelpark Arenberg 20, B-3001 Leuven, Belgium
| | - Kathleen Marchal
- Department of Information Technology, iGent Toren, Technologiepark 15, 9052 Gent, Belgium.,Department of Plant Biotechnology and Bioinformatics, UGent, Technologiepark 927, 9052 Gent, Belgium.,Bioinformatics Institute Ghent, Technologiepark 927, 9052 Gent, Belgium.,Department of Genetics, University of Pretoria, Hatfield Campus, Pretoria 0028, South Africa
| |
Collapse
|
783
|
Thiébaut R, Esmiol S, Lecine P, Mahfouz B, Hermant A, Nicoletti C, Parnis S, Perroy J, Borg JP, Pascoe L, Hugot JP, Ollendorff V. Characterization and Genetic Analyses of New Genes Coding for NOD2 Interacting Proteins. PLoS One 2016; 11:e0165420. [PMID: 27812135 PMCID: PMC5094585 DOI: 10.1371/journal.pone.0165420] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 10/11/2016] [Indexed: 01/26/2023] Open
Abstract
NOD2 contributes to the innate immune response and to the homeostasis of the intestinal mucosa. In response to its bacterial ligand, NOD2 interacts with RICK and activates the NF-κB and MAPK pathways, inducing gene transcription and synthesis of proteins required to initiate a balanced immune response. Mutations in NOD2 have been associated with an increased risk of Crohn’s Disease (CD), a disabling inflammatory bowel disease (IBD). Because NOD2 signaling plays a key role in CD, it is important to further characterize the network of protein interacting with NOD2. Using yeast two hybrid (Y2H) screens, we identified new NOD2 interacting proteins (NIP). The primary interaction was confirmed by coimmunoprecipitation and/or bioluminescence resonance energy transfer (BRET) experiments for 11 of these proteins (ANKHD1, CHMP5, SDCCAG3, TRIM41, LDOC1, PPP1R12C, DOCK7, VIM, KRT15, PPP2R3B, and C10Orf67). These proteins are involved in diverse functions, including endosomal sorting complexes required for transport (ESCRT), cytoskeletal architecture and signaling regulation. Additionally, we show that the interaction of 8 NIPs is compromised with the 3 main CD associated NOD2 mutants (R702W, G908R and 1007fs). Furthermore, to determine whether these NOD2 protein partners could be encoded by IBD susceptibility genes, a transmission disequilibrium test (TDT) was performed on 101 single nucleotide polymorphisms (SNPs) and the main corresponding haplotypes in genes coding for 15 NIPs using a set of 343 IBD families with 556 patients. Overall this work did not increase the number of IBD susceptibility genes but extends the NOD2 protein interaction network and suggests that NOD2 interactome and signaling depend upon the NOD2 mutation profile in CD.
Collapse
Affiliation(s)
- Raphaële Thiébaut
- UMR1149, INSERM et Université Paris Diderot-Sorbonne Paris-Cité, 75018, Paris, France
| | - Sophie Esmiol
- INRA, UMR866, DMEM, Université de Montpellier, Montpellier, France
| | - Patrick Lecine
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, "Cell Polarity, Cell signaling and Cancer - Equipe labellisée Ligue Contre le Cancer", Marseille, France
| | - Batoul Mahfouz
- UMR1149, INSERM et Université Paris Diderot-Sorbonne Paris-Cité, 75018, Paris, France
| | - Aurelie Hermant
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, "Cell Polarity, Cell signaling and Cancer - Equipe labellisée Ligue Contre le Cancer", Marseille, France
| | - Cendrine Nicoletti
- Aix Marseille Université, Centrale Marseille, CNRS, ISM2 UMR7313, 13397, Marseille, France
| | - Stephane Parnis
- Aix Marseille Université, Centrale Marseille, CNRS, ISM2 UMR7313, 13397, Marseille, France
| | - Julie Perroy
- CNRS, UMR-5203, Institut de Génomique Fonctionnelle, Montpellier, F-34094, France
- INSERM, U1191, Montpellier, F-34094, France
- Université de Montpellier, UMR-5203, Montpellier, F-34094, France
| | - Jean-Paul Borg
- Aix Marseille Univ, CNRS, INSERM, Institut Paoli-Calmettes, CRCM, "Cell Polarity, Cell signaling and Cancer - Equipe labellisée Ligue Contre le Cancer", Marseille, France
| | | | - Jean-Pierre Hugot
- UMR1149, INSERM et Université Paris Diderot-Sorbonne Paris-Cité, 75018, Paris, France
- Assistance Publique Hôpitaux de Paris, service de gastroentérologie pédiatrique, Hôpital Robert Debré, 75019, Paris, France
| | - Vincent Ollendorff
- INRA, UMR866, DMEM, Université de Montpellier, Montpellier, France
- * E-mail:
| |
Collapse
|
784
|
Kennedy RB, Ovsyannikova IG, Haralambieva IH, Oberg AL, Zimmermann MT, Grill DE, Poland GA. Immunosenescence-Related Transcriptomic and Immunologic Changes in Older Individuals Following Influenza Vaccination. Front Immunol 2016; 7:450. [PMID: 27853459 PMCID: PMC5089977 DOI: 10.3389/fimmu.2016.00450] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Accepted: 10/10/2016] [Indexed: 12/24/2022] Open
Abstract
The goal of annual influenza vaccination is to reduce mortality and morbidity associated with this disease through the generation of protective immune responses. The objective of the current study was to examine markers of immunosenescence and identify immunosenescence-related differences in gene expression, gene regulation, cytokine secretion, and immunologic changes in an older study population receiving seasonal influenza A/H1N1 vaccination. Surprisingly, prior studies in this cohort revealed weak correlations between immunosenescence markers and humoral immune response to vaccination. In this report, we further examined the relationship of each immunosenescence marker (age, T cell receptor excision circle frequency, telomerase expression, percentage of CD28− CD4+ T cells, percentage of CD28− CD8+ T cells, and the CD4/CD8 T cell ratio) with additional markers of immune response (serum cytokine and chemokine expression) and measures of gene expression and/or regulation. Many of the immunosenescence markers indeed correlated with distinct sets of individual DNA methylation sites, miRNA expression levels, mRNA expression levels, serum cytokines, and leukocyte subsets. However, when the individual immunosenescence markers were grouped by pathways or functional terms, several shared biological functions were identified: antigen processing and presentation pathways, MAPK, mTOR, TCR, BCR, and calcium signaling pathways, as well as key cellular metabolic, proliferation and survival activities. Furthermore, the percent of CD4+ and/or CD8+ T cells lacking CD28 expression also correlated with miRNAs regulating clusters of genes known to be involved in viral infection. Integrated (DNA methylation, mRNA, miRNA, and protein levels) network biology analysis of immunosenescence-related pathways and genesets identified both known pathways (e.g., chemokine signaling, CTL, and NK cell activity), as well as a gene expression module not previously annotated with a known function. These results may improve our ability to predict immune responses to influenza and aid in new vaccine development, and highlight the need for additional studies to better define and characterize immunosenescence.
Collapse
Affiliation(s)
- Richard B Kennedy
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic , Rochester, MN , USA
| | - Inna G Ovsyannikova
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic , Rochester, MN , USA
| | - Iana H Haralambieva
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic , Rochester, MN , USA
| | - Ann L Oberg
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic , Rochester, MN , USA
| | - Michael T Zimmermann
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic , Rochester, MN , USA
| | - Diane E Grill
- Department of Health Sciences Research, Division of Biomedical Statistics and Informatics, Mayo Clinic , Rochester, MN , USA
| | - Gregory A Poland
- Mayo Clinic Vaccine Research Group, Department of General Internal Medicine, Mayo Clinic , Rochester, MN , USA
| |
Collapse
|
785
|
Vasconcellos R, Alvarenga ÉC, Parreira RC, Lima SS, Resende RR. Exploring the cell signalling in hepatocyte differentiation. Cell Signal 2016; 28:1773-88. [DOI: 10.1016/j.cellsig.2016.08.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2016] [Revised: 08/18/2016] [Accepted: 08/18/2016] [Indexed: 02/08/2023]
|
786
|
Lievens S, Van der Heyden J, Masschaele D, De Ceuninck L, Petta I, Gupta S, De Puysseleyr V, Vauthier V, Lemmens I, De Clercq DJH, Defever D, Vanderroost N, De Smet AS, Eyckerman S, Van Calenbergh S, Martens L, De Bosscher K, Libert C, Hill DE, Vidal M, Tavernier J. Proteome-scale Binary Interactomics in Human Cells. Mol Cell Proteomics 2016; 15:3624-3639. [PMID: 27803151 DOI: 10.1074/mcp.m116.061994] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 10/23/2016] [Indexed: 12/11/2022] Open
Abstract
Because proteins are the main mediators of most cellular processes they are also prime therapeutic targets. Identifying physical links among proteins and between drugs and their protein targets is essential in order to understand the mechanisms through which both proteins themselves and the molecules they are targeted with act. Thus, there is a strong need for sensitive methods that enable mapping out these biomolecular interactions. Here we present a robust and sensitive approach to screen proteome-scale collections of proteins for binding to proteins or small molecules using the well validated MAPPIT (Mammalian Protein-Protein Interaction Trap) and MASPIT (Mammalian Small Molecule-Protein Interaction Trap) assays. Using high-density reverse transfected cell microarrays, a close to proteome-wide collection of human ORF clones can be screened for interactors at high throughput. The versatility of the platform is demonstrated through several examples. With MAPPIT, we screened a 15k ORF library for binding partners of RNF41, an E3 ubiquitin protein ligase implicated in receptor sorting, identifying known and novel interacting proteins. The potential related to the fact that MAPPIT operates in living human cells is illustrated in a screen where the protein collection is scanned for interactions with the glucocorticoid receptor (GR) in its unliganded versus dexamethasone-induced activated state. Several proteins were identified the interaction of which is modulated upon ligand binding to the GR, including a number of previously reported GR interactors. Finally, the screening technology also enables detecting small molecule target proteins, which in many drug discovery programs represents an important hurdle. We show the efficiency of MASPIT-based target profiling through screening with tamoxifen, a first-line breast cancer drug, and reversine, an investigational drug with interesting dedifferentiation and antitumor activity. In both cases, cell microarray screens yielded known and new potential drug targets highlighting the utility of the technology beyond fundamental biology.
Collapse
Affiliation(s)
- Sam Lievens
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - José Van der Heyden
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Delphine Masschaele
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Leentje De Ceuninck
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Ioanna Petta
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium.,‖Inflammation Research Center, VIB, Ghent, Belgium.,**Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - Surya Gupta
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Veronic De Puysseleyr
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Virginie Vauthier
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Irma Lemmens
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | | | - Dieter Defever
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Nele Vanderroost
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Anne-Sophie De Smet
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Sven Eyckerman
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | | | - Lennart Martens
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Karolien De Bosscher
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium.,§Department of Biochemistry, Ghent University, Ghent, Belgium
| | - Claude Libert
- ‖Inflammation Research Center, VIB, Ghent, Belgium.,**Department of Biomedical Molecular Biology, Ghent University, Ghent, Belgium
| | - David E Hill
- ‡‡Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,§§Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Marc Vidal
- ‡‡Center for Cancer Systems Biology (CCSB) and Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts.,§§Department of Genetics, Harvard Medical School, Boston, Massachusetts
| | - Jan Tavernier
- From the ‡Medical Biotechnology Center, VIB, Ghent, Belgium; .,§Department of Biochemistry, Ghent University, Ghent, Belgium
| |
Collapse
|
787
|
An Integrative Analysis of Preeclampsia Based on the Construction of an Extended Composite Network Featuring Protein-Protein Physical Interactions and Transcriptional Relationships. PLoS One 2016; 11:e0165849. [PMID: 27802351 PMCID: PMC5089765 DOI: 10.1371/journal.pone.0165849] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2016] [Accepted: 10/18/2016] [Indexed: 11/19/2022] Open
Abstract
Preeclampsia (PE) is a pregnancy disorder defined by hypertension and proteinuria. This disease remains a major cause of maternal and fetal morbidity and mortality. Defective placentation is generally described as being at the root of the disease. The characterization of the transcriptome signature of the preeclamptic placenta has allowed to identify differentially expressed genes (DEGs). However, we still lack a detailed knowledge on how these DEGs impact the function of the placenta. The tools of network biology offer a methodology to explore complex diseases at a systems level. In this study we performed a cross-platform meta-analysis of seven publically available gene expression datasets comparing non-pathological and preeclamptic placentas. Using the rank product algorithm we identified a total of 369 DEGs consistently modified in PE. The DEGs were used as seeds to build both an extended physical protein-protein interactions network and a transcription factors regulatory network. Topological and clustering analysis was conducted to analyze the connectivity properties of the networks. Finally both networks were merged into a composite network which presents an integrated view of the regulatory pathways involved in preeclampsia and the crosstalk between them. This network is a useful tool to explore the relationship between the DEGs and enable hypothesis generation for functional experimentation.
Collapse
|
788
|
Ma J, Zhang X, Feng Y, Zhang H, Wang X, Zheng Y, Qiao W, Liu X. Structural and Functional Study of Apoptosis-linked Gene-2·Heme-binding Protein 2 Interactions in HIV-1 Production. J Biol Chem 2016; 291:26670-26685. [PMID: 27784779 DOI: 10.1074/jbc.m116.752444] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 10/01/2016] [Indexed: 01/10/2023] Open
Abstract
In the HIV-1 replication cycle, the endosomal sorting complex required for transport (ESCRT) machinery promotes viral budding and release in the late stages. In this process, the ESCRT proteins, ALIX and TSG101, are recruited through interactions with HIV-1 Gag p6. ALG-2, also known as PDCD6, interacts with both ALIX and TSG101 and bridges ESCRT-III and ESCRT-I. In this study, we show that ALG-2 affects HIV-1 production negatively at both the exogenous and endogenous levels. Through a yeast two-hybrid screen, we identified HEBP2 as the binding partner of ALG-2, and we solved the crystal structure of the ALG-2·HEBP2 complex. The function of ALG-2·HEBP2 complex in HIV-1 replication was further explored. ALG-2 inhibits HIV-1 production by affecting Gag expression and distribution, and HEBP2 might aid this process by tethering ALG-2 in the cytoplasm.
Collapse
Affiliation(s)
- Jing Ma
- From the State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071.,the Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xianfeng Zhang
- the CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, and
| | - Yanbin Feng
- From the State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071
| | - Hui Zhang
- From the State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071
| | - Xiaojun Wang
- the CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, and
| | - Yonghui Zheng
- the CAAS-Michigan State University Joint Laboratory of Innate Immunity, State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin 150069, and
| | - Wentao Qiao
- From the State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071, .,the Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Xinqi Liu
- From the State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin 300071,
| |
Collapse
|
789
|
Choi ES, Lee H, Lee CH, Goh SH. Overexpression of KLHL23 protein from read-through transcription of PHOSPHO2-KLHL23 in gastric cancer increases cell proliferation. FEBS Open Bio 2016; 6:1155-1164. [PMID: 27833855 PMCID: PMC5095152 DOI: 10.1002/2211-5463.12136] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2016] [Revised: 09/20/2016] [Accepted: 09/28/2016] [Indexed: 01/05/2023] Open
Abstract
Gene fusion, as a prototypical pathognomonic mutation, contributes to genome complexity, and the cis‐transcription‐induced gene fusions generated by read‐through transcription of adjacent genes have been found to be important for tumor development. We screened read‐through transcription events from stomach adenocarcinoma RNA‐seq data and selected three candidates PHOSPHO2‐KLHL23, RPL17‐C18orf32, and PRR5‐ARHGAP8, to assess their biological role in gastric cancer. The expression of all three read‐through fusion transcripts was confirmed in gastric cancer cell lines and paired normal/tumor gastric cancer tissues by real‐time quantitative reverse transcription polymerase chain reaction and their expression was found to be significantly higher in the tumor (P < 0.05; n = 75). The correlation between the expression level and clinicopathological information was statistically analyzed. The level of the PHOSPHO2‐KLHL23 read‐through fusion transcript correlated with the Lauren classification and was significantly associated with the presence of perineural invasion. Overexpression of KLHL23 from PHOSPHO2‐KLHL23 read‐through transcript led to a significant increase in cell proliferation and resistance to anticancer drug treatment. Silencing of KLHL23 expression decreased cyclin D1 levels. The expression of KLHL23 from prevalent read‐through transcripts of PHOSPHO2‐KLHL23 in gastric cancer may undermine the efficacy of anticancer drug treatment.
Collapse
Affiliation(s)
- Eun-Seok Choi
- Precision Medicine Branch Research Institute National Cancer Center Goyang Gyeonggi-do Korea; Department of Environmental Medical Biology Institute of Tropical Medicine Yonsei University College of Medicine Seoul Korea
| | - Hanna Lee
- Precision Medicine Branch Research Institute National Cancer Center Goyang Gyeonggi-do Korea
| | - Chang-Hun Lee
- Cancer Cell and Molecular Biology Branch Research Institute National Cancer Center Goyang Gyeonggi-do Korea
| | - Sung-Ho Goh
- Precision Medicine Branch Research Institute National Cancer Center Goyang Gyeonggi-do Korea
| |
Collapse
|
790
|
Garzón JI, Deng L, Murray D, Shapira S, Petrey D, Honig B. A computational interactome and functional annotation for the human proteome. eLife 2016; 5. [PMID: 27770567 PMCID: PMC5115866 DOI: 10.7554/elife.18715] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2016] [Accepted: 10/19/2016] [Indexed: 12/14/2022] Open
Abstract
We present a database, PrePPI (Predicting Protein-Protein Interactions), of more than 1.35 million predicted protein-protein interactions (PPIs). Of these at least 127,000 are expected to constitute direct physical interactions although the actual number may be much larger (~500,000). The current PrePPI, which contains predicted interactions for about 85% of the human proteome, is related to an earlier version but is based on additional sources of interaction evidence and is far larger in scope. The use of structural relationships allows PrePPI to infer numerous previously unreported interactions. PrePPI has been subjected to a series of validation tests including reproducing known interactions, recapitulating multi-protein complexes, analysis of disease associated SNPs, and identifying functional relationships between interacting proteins. We show, using Gene Set Enrichment Analysis (GSEA), that predicted interaction partners can be used to annotate a protein's function. We provide annotations for most human proteins, including many annotated as having unknown function.
Collapse
Affiliation(s)
- José Ignacio Garzón
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, United States
| | - Lei Deng
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, United States.,School of Software, Central South University, Changsha, China
| | - Diana Murray
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, United States
| | - Sagi Shapira
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, United States.,Department of Microbiology and Immunology, Columbia University, New York, United States
| | - Donald Petrey
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, United States.,Howard Hughes Medical Institute, Columbia University, New York, United States
| | - Barry Honig
- Center for Computational Biology and Bioinformatics, Department of Systems Biology, Columbia University, New York, United States.,Howard Hughes Medical Institute, Columbia University, New York, United States.,Department of Biochemistry and Molecular Biophysics, Columbia University, New York, United States.,Department of Medicine, Columbia University, New York, United States.,Zuckerman Mind Brain Behavior Institute, Columbia University, New York, United States
| |
Collapse
|
791
|
Interactome-transcriptome analysis discovers signatures complementary to GWAS Loci of Type 2 Diabetes. Sci Rep 2016; 6:35228. [PMID: 27752041 PMCID: PMC5067504 DOI: 10.1038/srep35228] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Accepted: 09/26/2016] [Indexed: 01/13/2023] Open
Abstract
Protein interactions play significant roles in complex diseases. We analyzed peripheral blood mononuclear cells (PBMC) transcriptome using a multi-method strategy. We constructed a tissue-specific interactome (T2Di) and identified 420 molecular signatures associated with T2D-related comorbidity and symptoms, mainly implicated in inflammation, adipogenesis, protein phosphorylation and hormonal secretion. Apart from explaining the residual associations within the DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) study, the T2Di signatures were enriched in pathogenic cell type-specific regulatory elements related to fetal development, immunity and expression quantitative trait loci (eQTL). The T2Di revealed a novel locus near a well-established GWAS loci AChE, in which SRRT interacts with JAZF1, a T2D-GWAS gene implicated in pancreatic function. The T2Di also included known anti-diabetic drug targets (e.g. PPARD, MAOB) and identified possible druggable targets (e.g. NCOR2, PDGFR). These T2Di signatures were validated by an independent computational method, and by expression data of pancreatic islet, muscle and liver with some of the signatures (CEBPB, SREBF1, MLST8, SRF, SRRT and SLC12A9) confirmed in PBMC from an independent cohort of 66 T2D and 66 control subjects. By combining prior knowledge and transcriptome analysis, we have constructed an interactome to explain the multi-layered regulatory pathways in T2D.
Collapse
|
792
|
Abstract
Genes carrying mutations associated with genetic diseases are present in all human cells; yet, clinical manifestations of genetic diseases are usually highly tissue-specific. Although some disease genes are expressed only in selected tissues, the expression patterns of disease genes alone cannot explain the observed tissue specificity of human diseases. Here we hypothesize that for a disease to manifest itself in a particular tissue, a whole functional subnetwork of genes (disease module) needs to be expressed in that tissue. Driven by this hypothesis, we conducted a systematic study of the expression patterns of disease genes within the human interactome. We find that genes expressed in a specific tissue tend to be localized in the same neighborhood of the interactome. By contrast, genes expressed in different tissues are segregated in distinct network neighborhoods. Most important, we show that it is the integrity and the completeness of the expression of the disease module that determines disease manifestation in selected tissues. This approach allows us to construct a disease-tissue network that confirms known and predicts unexpected disease-tissue associations.
Collapse
|
793
|
Network diffusion-based analysis of high-throughput data for the detection of differentially enriched modules. Sci Rep 2016; 6:34841. [PMID: 27731320 PMCID: PMC5059623 DOI: 10.1038/srep34841] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 08/19/2016] [Indexed: 11/08/2022] Open
Abstract
A relation exists between network proximity of molecular entities in interaction networks, functional similarity and association with diseases. The identification of network regions associated with biological functions and pathologies is a major goal in systems biology. We describe a network diffusion-based pipeline for the interpretation of different types of omics in the context of molecular interaction networks. We introduce the network smoothing index, a network-based quantity that allows to jointly quantify the amount of omics information in genes and in their network neighbourhood, using network diffusion to define network proximity. The approach is applicable to both descriptive and inferential statistics calculated on omics data. We also show that network resampling, applied to gene lists ranked by quantities derived from the network smoothing index, indicates the presence of significantly connected genes. As a proof of principle, we identified gene modules enriched in somatic mutations and transcriptional variations observed in samples of prostate adenocarcinoma (PRAD). In line with the local hypothesis, network smoothing index and network resampling underlined the existence of a connected component of genes harbouring molecular alterations in PRAD.
Collapse
|
794
|
Chromatin structure-based prediction of recurrent noncoding mutations in cancer. Nat Genet 2016; 48:1321-1326. [PMID: 27723759 DOI: 10.1038/ng.3682] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 08/29/2016] [Indexed: 12/15/2022]
Abstract
Recurrence is a hallmark of cancer-driving mutations. Recurrent mutations can arise at the same site or affect the same gene at different sites. Here we identified a set of mutations arising in individual samples and altering different cis-regulatory elements that converge on a common gene via chromatin interactions. The mutations and genes identified in this fashion showed strong relevance to cancer, in contrast to noncoding mutations with site-specific recurrence only. We developed a prediction method that identifies potentially recurrent mutations on the basis of the features shared by mutations whose recurrence is observed in a given cohort. Our method was capable of accurately predicting recurrent mutations at the level of target genes but not mutations recurring at the same site. We experimentally validated predicted mutations in distal regulatory regions of the TERT gene. In conclusion, we propose a novel approach to discovering potential cancer-driving mutations in noncoding regions.
Collapse
|
795
|
|
796
|
Iñigo S, Durand AN, Ritter A, Le Gall S, Termathe M, Klassen R, Tohge T, De Coninck B, Van Leene J, De Clercq R, Cammue BPA, Fernie AR, Gevaert K, De Jaeger G, Leidel SA, Schaffrath R, Van Lijsebettens M, Pauwels L, Goossens A. Glutaredoxin GRXS17 Associates with the Cytosolic Iron-Sulfur Cluster Assembly Pathway. PLANT PHYSIOLOGY 2016; 172:858-873. [PMID: 27503603 PMCID: PMC5047072 DOI: 10.1104/pp.16.00261] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Accepted: 08/03/2016] [Indexed: 05/12/2023]
Abstract
Cytosolic monothiol glutaredoxins (GRXs) are required in iron-sulfur (Fe-S) cluster delivery and iron sensing in yeast and mammals. In plants, it is unclear whether they have similar functions. Arabidopsis (Arabidopsis thaliana) has a sole class II cytosolic monothiol GRX encoded by GRXS17 Here, we used tandem affinity purification to establish that Arabidopsis GRXS17 associates with most known cytosolic Fe-S assembly (CIA) components. Similar to mutant plants with defective CIA components, grxs17 loss-of-function mutants showed some degree of hypersensitivity to DNA damage and elevated expression of DNA damage marker genes. We also found that several putative Fe-S client proteins directly bind to GRXS17, such as XANTHINE DEHYDROGENASE1 (XDH1), involved in the purine salvage pathway, and CYTOSOLIC THIOURIDYLASE SUBUNIT1 and CYTOSOLIC THIOURIDYLASE SUBUNIT2, both essential for the 2-thiolation step of 5-methoxycarbonylmethyl-2-thiouridine (mcm5s2U) modification of tRNAs. Correspondingly, profiling of the grxs17-1 mutant pointed to a perturbed flux through the purine degradation pathway and revealed that it phenocopied mutants in the elongator subunit ELO3, essential for the mcm5 tRNA modification step, although we did not find XDH1 activity or tRNA thiolation to be markedly reduced in the grxs17-1 mutant. Taken together, our data suggest that plant cytosolic monothiol GRXs associate with the CIA complex, as in other eukaryotes, and contribute to, but are not essential for, the correct functioning of client Fe-S proteins in unchallenged conditions.
Collapse
Affiliation(s)
- Sabrina Iñigo
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Astrid Nagels Durand
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Andrés Ritter
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Sabine Le Gall
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Martin Termathe
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Roland Klassen
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Takayuki Tohge
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Barbara De Coninck
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Jelle Van Leene
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Rebecca De Clercq
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Bruno P A Cammue
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Alisdair R Fernie
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Kris Gevaert
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Geert De Jaeger
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Sebastian A Leidel
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Raffael Schaffrath
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Mieke Van Lijsebettens
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Laurens Pauwels
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| | - Alain Goossens
- Department of Plant Systems Biology, VIB, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., B.D.C., J.V.L., R.D.C., B.P.A.C., G.D.J., M.V.L., L.P., A.G.);Department of Plant Biotechnology and Bioinformatics, Ghent University, B-9052 Ghent, Belgium (S.I., A.N.D., A.R., S.L.G., J.V.L., R.D.C., G.D.J., M.V.L., L.P., A.G.);Max Planck Research Group for RNA Biology, Max Planck Institute for Molecular Biomedicine, 48149 Muenster, Germany (M.T., S.A.L.);Institut für Biologie, Fachgebiet Mikrobiologie, Universität Kassel, D-34132 Kassel, Germany (R.K., R.S.);Max Planck Institute of Molecular Plant Physiology, D-14476 Potsdam-Golm, Germany (T.T., A.R.F.);Centre of Microbial and Plant Genetics, Katholieke Universiteit Leuven, B-3001 Leuven, Belgium (B.D.C., B.P.A.C.);Cells-in-Motion Cluster of Excellence (M.T., S.A.L.) and Faculty of Medicine (S.A.L.), University of Muenster, 48149 Muenster, Germany;Department of Medical Protein Research, VIB, B-9000 Ghent, Belgium (K.G.); andDepartment of Biochemistry, Ghent University, B-9000 Ghent, Belgium (K.G.)
| |
Collapse
|
797
|
Eyckerman S, Impens F, Van Quickelberghe E, Samyn N, Vandemoortele G, De Sutter D, Tavernier J, Gevaert K. Intelligent Mixing of Proteomes for Elimination of False Positives in Affinity Purification-Mass Spectrometry. J Proteome Res 2016; 15:3929-3937. [PMID: 27640904 DOI: 10.1021/acs.jproteome.6b00517] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Protein complexes are essential in all organizational and functional aspects of the cell. Different strategies currently exist for analyzing such protein complexes by mass spectrometry, including affinity purification (AP-MS) and proximal labeling-based strategies. However, the high sensitivity of current mass spectrometers typically results in extensive protein lists mainly consisting of nonspecifically copurified proteins. Finding the true positive interactors in these lists remains highly challenging. Here, we report a powerful design based on differential labeling with stable isotopes combined with nonequal mixing of control and experimental samples to discover bona fide interaction partners in AP-MS experiments. We apply this intelligent mixing of proteomes (iMixPro) concept to overexpression experiments for RAF1, RNF41, and TANK and also to engineered cell lines expressing epitope-tagged endogenous PTPN14, JIP3, and IQGAP1. For all baits, we confirmed known interactions and found a number of novel interactions. The results for RNF41 and TANK were compared to a classical affinity purification experiment, which demonstrated the efficiency and specificity of the iMixPro approach.
Collapse
Affiliation(s)
- Sven Eyckerman
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Francis Impens
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,VIB Proteomics Expertise Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Emmy Van Quickelberghe
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Noortje Samyn
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Giel Vandemoortele
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Delphine De Sutter
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Jan Tavernier
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| | - Kris Gevaert
- VIB Medical Biotechnology Center , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium.,Department of Biochemistry, Ghent University , Albert Baertsoenkaai 3, B-9000 Ghent, Belgium
| |
Collapse
|
798
|
Tonddast-Navaei S, Skolnick J. Are protein-protein interfaces special regions on a protein's surface? J Chem Phys 2016; 143:243149. [PMID: 26723634 DOI: 10.1063/1.4937428] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Protein-protein interactions (PPIs) are involved in many cellular processes. Experimentally obtained protein quaternary structures provide the location of protein-protein interfaces, the surface region of a given protein that interacts with another. These regions are termed half-interfaces (HIs). Canonical HIs cover roughly one third of a protein's surface and were found to have more hydrophobic residues than the non-interface surface region. In addition, the classical view of protein HIs was that there are a few (if not one) HIs per protein that are structurally and chemically unique. However, on average, a given protein interacts with at least a dozen others. This raises the question of whether they use the same or other HIs. By copying HIs from monomers with the same folds in solved quaternary structures, we introduce the concept of geometric HIs (HIs whose geometry has a significant match to other known interfaces) and show that on average they cover three quarters of a protein's surface. We then demonstrate that in some cases, these geometric HI could result in real physical interactions (which may or may not be biologically relevant). The composition of the new HIs is on average more charged compared to most known ones, suggesting that the current protein interface database is biased towards more hydrophobic, possibly more obligate, complexes. Finally, our results provide evidence for interface fuzziness and PPI promiscuity. Thus, the classical view of unique, well defined HIs needs to be revisited as HIs are another example of coarse-graining that is used by nature.
Collapse
Affiliation(s)
- Sam Tonddast-Navaei
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 250 14th Street N.W., Atlanta, Georgia 30318, USA
| | - Jeffrey Skolnick
- Center for the Study of Systems Biology, School of Biology, Georgia Institute of Technology, 250 14th Street N.W., Atlanta, Georgia 30318, USA
| |
Collapse
|
799
|
Vuorinen EM, Rajala NK, Rauhala HE, Nurminen AT, Hytönen VP, Kallioniemi A. Search for KPNA7 cargo proteins in human cells reveals MVP and ZNF414 as novel regulators of cancer cell growth. Biochim Biophys Acta Mol Basis Dis 2016; 1863:211-219. [PMID: 27664836 DOI: 10.1016/j.bbadis.2016.09.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 08/26/2016] [Accepted: 09/20/2016] [Indexed: 12/20/2022]
Abstract
Karyopherin alpha 7 (KPNA7) belongs to a family of nuclear import proteins that recognize and bind nuclear localization signals (NLSs) in proteins to be transported to the nucleus. Previously we found that KPNA7 is overexpressed in a subset of pancreatic cancer cell lines and acts as a critical regulator of growth in these cells. This characteristic of KPNA7 is likely to be mediated by its cargo proteins that are still mainly unknown. Here, we used protein affinity chromatography in Hs700T and MIA PaCa-2 pancreatic cancer cell lines and identified 377 putative KPNA7 cargo proteins, most of which were known or predicted to localize to the nucleus. The interaction was confirmed for two of the candidates, MVP and ZNF414, using co-immunoprecipitation, and their transport to the nucleus was hindered by siRNA based KPNA7 silencing. Most importantly, silencing of MVP and ZNF414 resulted in marked reduction in Hs700T cell growth. In conclusion, these data uncover two previously unknown human KPNA7 cargo proteins with distinct roles as novel regulators of pancreatic cancer cell growth, thus deepening our understanding on the contribution of nuclear transport in cancer pathogenesis.
Collapse
Affiliation(s)
- Elisa M Vuorinen
- University of Tampere, BioMediTech, PL 100, 33014 TAMPEREEN YLIOPISTO, Tampere, Finland; Fimlab laboratories, Biokatu 4, 33520 Tampere, Finland.
| | - Nina K Rajala
- University of Tampere, BioMediTech, PL 100, 33014 TAMPEREEN YLIOPISTO, Tampere, Finland; Fimlab laboratories, Biokatu 4, 33520 Tampere, Finland.
| | - Hanna E Rauhala
- University of Tampere, BioMediTech, PL 100, 33014 TAMPEREEN YLIOPISTO, Tampere, Finland.
| | - Anssi T Nurminen
- University of Tampere, BioMediTech, PL 100, 33014 TAMPEREEN YLIOPISTO, Tampere, Finland; Fimlab laboratories, Biokatu 4, 33520 Tampere, Finland.
| | - Vesa P Hytönen
- University of Tampere, BioMediTech, PL 100, 33014 TAMPEREEN YLIOPISTO, Tampere, Finland; Fimlab laboratories, Biokatu 4, 33520 Tampere, Finland.
| | - Anne Kallioniemi
- University of Tampere, BioMediTech, PL 100, 33014 TAMPEREEN YLIOPISTO, Tampere, Finland; Fimlab laboratories, Biokatu 4, 33520 Tampere, Finland.
| |
Collapse
|
800
|
Abstract
A clear definition of what constitutes “Big Data” is difficult to identify, but we find it most useful to define Big Data as a data collection that is complete. By this criterion, researchers on Caenorhabditis elegans have a long history of collecting Big Data, since the organism was selected with the idea of obtaining a complete biological description and understanding of development. The complete wiring diagram of the nervous system, the complete cell lineage, and the complete genome sequence provide a framework to phrase and test hypotheses. Given this history, it might be surprising that the number of “complete” data sets for this organism is actually rather small—not because of lack of effort, but because most types of biological experiments are not currently amenable to complete large-scale data collection. Many are also not inherently limited, so that it becomes difficult to even define completeness. At present, we only have partial data on mutated genes and their phenotypes, gene expression, and protein–protein interaction—important data for many biological questions. Big Data can point toward unexpected correlations, and these unexpected correlations can lead to novel investigations; however, Big Data cannot establish causation. As a result, there is much excitement about Big Data, but there is also a discussion on just what Big Data contributes to solving a biological problem. Because of its relative simplicity, C. elegans is an ideal test bed to explore this issue and at the same time determine what is necessary to build a multicellular organism from a single cell.
Collapse
Affiliation(s)
- Harald Hutter
- Department of Biological Sciences, Simon Fraser University, Burnaby, BC V5A 1S6, Canada
| | - Donald Moerman
- Department of Zoology, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| |
Collapse
|